Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Rev Neurosci ; 19(4): 235-249, 2018 04.
Article in English | MEDLINE | ID: mdl-29515192

ABSTRACT

Lactate in the brain has long been associated with ischaemia; however, more recent evidence shows that it can be found there under physiological conditions. In the brain, lactate is formed predominantly in astrocytes from glucose or glycogen in response to neuronal activity signals. Thus, neurons and astrocytes show tight metabolic coupling. Lactate is transferred from astrocytes to neurons to match the neuronal energetic needs, and to provide signals that modulate neuronal functions, including excitability, plasticity and memory consolidation. In addition, lactate affects several homeostatic functions. Overall, lactate ensures adequate energy supply, modulates neuronal excitability levels and regulates adaptive functions in order to set the 'homeostatic tone' of the nervous system.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Energy Metabolism , Lactic Acid/metabolism , Neurons/metabolism , Animals , Humans , Memory/physiology , Neuronal Plasticity , Signal Transduction
2.
PLoS One ; 10(10): e0141568, 2015.
Article in English | MEDLINE | ID: mdl-26513352

ABSTRACT

We examined the expression of genes related to brain energy metabolism and particularly those encoding glia (astrocyte)-specific functions in the dorsal hippocampus subsequent to learning. Context-dependent avoidance behavior was tested in mice using the step-through Inhibitory Avoidance (IA) paradigm. Animals were sacrificed 3, 9, 24, or 72 hours after training or 3 hours after retention testing. The quantitative determination of mRNA levels revealed learning-induced changes in the expression of genes thought to be involved in astrocyte-neuron metabolic coupling in a time dependent manner. Twenty four hours following IA training, an enhanced gene expression was seen, particularly for genes encoding monocarboxylate transporters 1 and 4 (MCT1, MCT4), alpha2 subunit of the Na/K-ATPase and glucose transporter type 1. To assess the functional role for one of these genes in learning, we studied MCT1 deficient mice and found that they exhibit impaired memory in the inhibitory avoidance task. Together, these observations indicate that neuron-glia metabolic coupling undergoes metabolic adaptations following learning as indicated by the change in expression of key metabolic genes.


Subject(s)
Astrocytes/metabolism , Avoidance Learning , Hippocampus/metabolism , Memory, Long-Term , Neurons/metabolism , RNA, Messenger/metabolism , Animals , Astrocytes/physiology , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Hippocampus/cytology , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Neurons/physiology , RNA, Messenger/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism
3.
Neuron ; 86(4): 883-901, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25996133

ABSTRACT

The energy demands of the brain are high: they account for at least 20% of the body's energy consumption. Evolutionary studies indicate that the emergence of higher cognitive functions in humans is associated with an increased glucose utilization and expression of energy metabolism genes. Functional brain imaging techniques such as fMRI and PET, which are widely used in human neuroscience studies, detect signals that monitor energy delivery and use in register with neuronal activity. Recent technological advances in metabolic studies with cellular resolution have afforded decisive insights into the understanding of the cellular and molecular bases of the coupling between neuronal activity and energy metabolism and point at a key role of neuron-astrocyte metabolic interactions. This article reviews some of the most salient features emerging from recent studies and aims at providing an integration of brain energy metabolism across resolution scales.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Energy Metabolism/physiology , Magnetic Resonance Imaging , Neurons/metabolism , Animals , Glucose/metabolism , Humans , Magnetic Resonance Imaging/methods
4.
PLoS Comput Biol ; 11(2): e1004036, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25719367

ABSTRACT

Glucose is the main energy substrate in the adult brain under normal conditions. Accumulating evidence, however, indicates that lactate produced in astrocytes (a type of glial cell) can also fuel neuronal activity. The quantitative aspects of this so-called astrocyte-neuron lactate shuttle (ANLS) are still debated. To address this question, we developed a detailed biophysical model of the brain's metabolic interactions. Our model integrates three modeling approaches, the Buxton-Wang model of vascular dynamics, the Hodgkin-Huxley formulation of neuronal membrane excitability and a biophysical model of metabolic pathways. This approach provides a template for large-scale simulations of the neuron-glia-vasculature (NGV) ensemble, and for the first time integrates the respective timescales at which energy metabolism and neuronal excitability occur. The model is constrained by relative neuronal and astrocytic oxygen and glucose utilization, by the concentration of metabolites at rest and by the temporal dynamics of NADH upon activation. These constraints produced four observations. First, a transfer of lactate from astrocytes to neurons emerged in response to activity. Second, constrained by activity-dependent NADH transients, neuronal oxidative metabolism increased first upon activation with a subsequent delayed astrocytic glycolysis increase. Third, the model correctly predicted the dynamics of extracellular lactate and oxygen as observed in vivo in rats. Fourth, the model correctly predicted the temporal dynamics of tissue lactate, of tissue glucose and oxygen consumption, and of the BOLD signal as reported in human studies. These findings not only support the ANLS hypothesis but also provide a quantitative mathematical description of the metabolic activation in neurons and glial cells, as well as of the macroscopic measurements obtained during brain imaging.


Subject(s)
Astrocytes/metabolism , Brain/blood supply , Brain/metabolism , Lactic Acid/metabolism , Models, Neurological , Neurons/metabolism , Animals , Cerebrovascular Circulation , Computational Biology , Computer Simulation , Extracellular Space , Glucose/metabolism , Humans , Models, Cardiovascular , NAD/metabolism , Oxygen Consumption , Rats , Sodium/metabolism
5.
Front Neurosci ; 9: 23, 2015.
Article in English | MEDLINE | ID: mdl-25709564

ABSTRACT

Glucose is the main energy substrate for the brain. There is now extensive evidence indicating that the metabolic profile of neural cells with regard to glucose utilization and glycolysis rate is not homogenous, with a marked propensity for glycolytic glucose processing in astrocytes compared to neurons. Methylglyoxal, a highly reactive dicarbonyl compound, is inevitably formed as a by-product of glycolysis. Methylglyoxal is a major cell-permeant precursor of advanced glycation end-products (AGEs), which are associated with several pathologies including diabetes, aging and neurodegenerative diseases. In normal situations, cells are protected against methylglyoxal toxicity by different mechanisms and in particular the glyoxalase system, which represents the most important pathway for the detoxification of methylglyoxal. While the neurotoxic effects of methylglyoxal and AGEs are well characterized, our understanding the glyoxalase system in the brain is more scattered. Considering the high energy requirements (i.e., glucose) of the brain, one should expect that the cerebral glyoxalase system is adequately fitted to handle methylglyoxal toxicity. This review focuses on our actual knowledge on the cellular aspects of the glyoxalase system in brain cells, in particular with regard to its activity in astrocytes and neurons. A main emerging concept is that these two neural cell types have different and energetically adapted glyoxalase defense mechanisms which may serve as protective mechanism against methylglyoxal-induced cellular damage.

6.
Metab Brain Dis ; 30(1): 263-79, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25399336

ABSTRACT

In 1995 Benington and Heller formulated an energy hypothesis of sleep centered on a key role of glycogen. It was postulated that a major function of sleep is to replenish glycogen stores in the brain that have been depleted during wakefulness which is associated to an increased energy demand. Astrocytic glycogen depletion participates to an increase of extracellular adenosine release which influences sleep homeostasis. Here, we will review some evidence obtained by studies addressing the question of a key role played by glycogen metabolism in sleep regulation as proposed by this hypothesis or by an alternative hypothesis named "glycogenetic" hypothesis as well as the importance of the confounding effect of glucocorticoïds. Even though actual collected data argue in favor of a role of sleep in brain energy balance-homeostasis, they do not support a critical and direct involvement of glycogen metabolism on sleep regulation. For instance, glycogen levels during the sleep-wake cycle are driven by different physiological signals and therefore appear more as a marker-integrator of brain energy status than a direct regulator of sleep homeostasis. In support of this we provide evidence that blockade of glycogen mobilization does not induce more sleep episodes during the active period while locomotor activity is reduced. These observations do not invalidate the energy hypothesis of sleep but indicate that underlying cellular mechanisms are more complex than postulated by Benington and Heller.


Subject(s)
Brain/metabolism , Energy Metabolism , Glycogen/metabolism , Homeostasis/physiology , Models, Neurological , Sleep/physiology , Adenosine/metabolism , Animals , Arousal/physiology , Astrocytes/metabolism , Citric Acid Cycle , Extracellular Fluid/metabolism , Glucocorticoids/physiology , Glucose/metabolism , Glycogenolysis , Glycolysis , Humans , Mice , Neurons/metabolism , Rats , Sleep Deprivation/metabolism
7.
Proc Natl Acad Sci U S A ; 111(33): 12228-33, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25071212

ABSTRACT

L-lactate is a product of aerobic glycolysis that can be used by neurons as an energy substrate. Here we report that in neurons L-lactate stimulates the expression of synaptic plasticity-related genes such as Arc, c-Fos, and Zif268 through a mechanism involving NMDA receptor activity and its downstream signaling cascade Erk1/2. L-lactate potentiates NMDA receptor-mediated currents and the ensuing increase in intracellular calcium. In parallel to this, L-lactate increases intracellular levels of NADH, thereby modulating the redox state of neurons. NADH mimics all of the effects of L-lactate on NMDA signaling, pointing to NADH increase as a primary mediator of L-lactate effects. The induction of plasticity genes is observed both in mouse primary neurons in culture and in vivo in the mouse sensory-motor cortex. These results provide insights for the understanding of the molecular mechanisms underlying the critical role of astrocyte-derived L-lactate in long-term memory and long-term potentiation in vivo. This set of data reveals a previously unidentified action of L-lactate as a signaling molecule for neuronal plasticity.


Subject(s)
Gene Expression/drug effects , Lactic Acid/pharmacology , N-Methylaspartate/metabolism , Neuronal Plasticity/genetics , Neurons/metabolism , Signal Transduction , Animals , Calcium/metabolism , Cells, Cultured , Mice
8.
Curr Drug Targets ; 14(11): 1308-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23469874

ABSTRACT

There is growing evidence that astrocytes are involved in the neuropathology of major depression. In particular, decreases in glial cell density observed in the cerebral cortex of individuals with major depressive disorder are accompanied by a reduction of several astrocytic markers suggesting that astrocyte dysfunction may contribute to the pathophysiology of major depression. In rodents, glial loss in the prefrontal cortex is sufficient to induce depressive-like behaviors and antidepressant treatment prevents the stress-induced reduction of astrocyte number in the hippocampus. Collectively, these data support the existence of a link between astrocyte loss or dysfunction, depressive-like behavior and antidepressant treatment. Astrocytes are increasingly recognized to play important roles in neuronal development, neurotransmission, synaptic plasticity and maintenance of brain homeostasis. It is also well established that astrocytes provide trophic, structural, and metabolic support to neurons. In this article, we review evidence that antidepressants regulate energy metabolism and neurotrophic factor expression with particular emphasis on studies in astrocytes. These observations support a role for astrocytes as new targets for antidepressants. The contribution of changes in astrocyte glucose metabolism and neurotrophic factor expression to the therapeutic effects of antidepressants remains to be established.


Subject(s)
Antidepressive Agents/therapeutic use , Astrocytes/drug effects , Depressive Disorder, Major/drug therapy , Energy Metabolism/drug effects , Nerve Growth Factors/metabolism , Stress, Psychological/drug therapy , Synaptic Transmission/drug effects , Animals , Antidepressive Agents/pharmacology , Astrocytes/metabolism , Depressive Disorder, Major/physiopathology , Disease Models, Animal , Gene Expression Regulation , Humans , Nerve Growth Factors/genetics , Rodentia , Stress, Psychological/physiopathology
9.
Cell Metab ; 14(6): 724-38, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22152301

ABSTRACT

The energy requirements of the brain are very high, and tight regulatory mechanisms operate to ensure adequate spatial and temporal delivery of energy substrates in register with neuronal activity. Astrocytes-a type of glial cell-have emerged as active players in brain energy delivery, production, utilization, and storage. Our understanding of neuroenergetics is rapidly evolving from a "neurocentric" view to a more integrated picture involving an intense cooperativity between astrocytes and neurons. This review focuses on the cellular aspects of brain energy metabolism, with a particular emphasis on the metabolic interactions between neurons and astrocytes.


Subject(s)
Astrocytes/metabolism , Brain/blood supply , Brain/physiology , Energy Metabolism/physiology , Models, Biological , Neurons/metabolism , Brain/cytology , Glycogen/metabolism , Lactic Acid/metabolism , Oxidative Stress/physiology , Regional Blood Flow/physiology
10.
J Neurosci ; 31(50): 18338-52, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22171037

ABSTRACT

The glyoxalase system is the most important pathway for the detoxification of methylglyoxal (MG), a highly reactive dicarbonyl compound mainly formed as a by-product of glycolysis. MG is a major precursor of advanced glycation end products (AGEs), which are associated with several neurodegenerative disorders. Although the neurotoxic effects of MG and AGEs are well characterized, little is known about the glyoxalase system in the brain, in particular with regards to its activity in different neural cell types. Results of the present study reveal that both enzymes composing the glyoxalase system [glyoxalase-1 (Glo-1) and Glo-2] were highly expressed in primary mouse astrocytes compared with neurons, which translated into higher enzymatic activity rates in astrocytes (9.9- and 2.5-fold, respectively). The presence of a highly efficient glyoxalase system in astrocytes was associated with lower accumulation of AGEs compared with neurons (as assessed by Western blotting), a sixfold greater resistance to MG toxicity, and the capacity to protect neurons against MG in a coculture system. In addition, Glo-1 downregulation using RNA interference strategies resulted in a loss of viability in neurons, but not in astrocytes. Finally, stimulation of neuronal glycolysis via lentiviral-mediated overexpression of 6-phosphofructose-2-kinase/fructose-2,6-bisphosphatase-3 resulted in increased MG levels and MG-modified proteins. Since MG is largely produced through glycolysis, this suggests that the poor capacity of neurons to upregulate their glycolytic flux as compared with astrocytes may be related to weaker defense mechanisms against MG toxicity. Accordingly, the neuroenergetic specialization taking place between these two cell types may serve as a protective mechanism against MG-induced neurotoxicity.


Subject(s)
Astrocytes/enzymology , Cytoprotection/physiology , Lactoylglutathione Lyase/metabolism , Neurons/enzymology , Thiolester Hydrolases/metabolism , Animals , Astrocytes/cytology , CHO Cells , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Cricetinae , Lactoylglutathione Lyase/genetics , Mice , Neurons/cytology , Thiolester Hydrolases/genetics
11.
PLoS One ; 6(7): e22875, 2011.
Article in English | MEDLINE | ID: mdl-21829542

ABSTRACT

Neurodegenerative and psychiatric disorders including Alzheimer's, Parkinson's or Huntington's diseases and schizophrenia have been associated with a deficit in glutathione (GSH). In particular, a polymorphism in the gene of glutamate cysteine ligase modulatory subunit (GCLM) is associated with schizophrenia. GSH is the most important intracellular antioxidant and is necessary for the removal of reactive by-products generated by the utilization of glucose for energy supply. Furthermore, glucose metabolism through the pentose phosphate pathway is a major source of NADPH, the cofactor necessary for the regeneration of reduced glutathione. This study aims at investigating glucose metabolism in cultured astrocytes from GCLM knockout mice, which show decreased GSH levels. No difference in the basal metabolism of glucose was observed between wild-type and knockout cells. In contrast, glycogen levels were lower and its turnover was higher in knockout astrocytes. These changes were accompanied by a decrease in the expression of the genes involved in its synthesis and degradation, including the protein targeting to glycogen. During an oxidative challenge induced by tert-Butylhydroperoxide, wild-type cells increased their glycogen mobilization and glucose uptake. However, knockout astrocytes were unable to mobilize glycogen following the same stress and they could increase their glucose utilization only following a major oxidative insult. Altogether, these results show that glucose metabolism and glycogen utilization are dysregulated in astrocytes showing a chronic deficit in GSH, suggesting that alterations of a fundamental aspect of brain energy metabolism is caused by GSH deficit and may therefore be relevant to metabolic dysfunctions observed in schizophrenia.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Glutamate-Cysteine Ligase/physiology , Glutathione/deficiency , Glycogen/metabolism , Oxidative Stress , Animals , Antioxidants/pharmacology , Blotting, Western , Carbon Dioxide/metabolism , Cells, Cultured , Chronic Disease , Energy Metabolism , Glucose/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/metabolism , Schizophrenia/pathology
12.
Psychopharmacology (Berl) ; 216(1): 75-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21301813

ABSTRACT

RATIONALE: The pharmacological actions of most antidepressants are ascribed to the modulation of serotonergic and/or noradrenergic transmission in the brain. During therapeutic treatment for major depression, fluoxetine, one of the most commonly prescribed selective serotonin reuptake inhibitor (SSRI) antidepressants, accumulates in the brain, suggesting that fluoxetine may interact with additional targets. In this context, there is increasing evidence that astrocytes are involved in the pathophysiology of major depression. OBJECTIVES: The aim of this study was to examine the effects of fluoxetine on the expression of neurotrophic/growth factors that have antidepressant properties and on glucose metabolism in cultured cortical astrocytes. RESULTS: Treatment of astrocytes with fluoxetine and paroxetine, another SSRI antidepressant, upregulated brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), and VGF mRNA expression. In contrast, the tricyclic antidepressants desipramine and imipramine did not affect the expression of these neurotrophic/growth factors. Analysis of the effects of fluoxetine on glucose metabolism revealed that fluoxetine reduces glycogen levels and increases glucose utilization and lactate release by astrocytes. Similar data were obtained with paroxetine, whereas imipramine and desipramine did not regulate glucose metabolism in this glial cell population. Our results also indicate that the effects of fluoxetine and paroxetine on glucose utilization, lactate release, and expression of BDNF, VEGF, and VGF are not mediated by serotonin-dependent mechanisms. CONCLUSIONS: These data suggest that, by increasing the expression of specific astrocyte-derived neurotrophic factors and lactate release from astrocytes, fluoxetine may contribute to normalize the trophic and metabolic support to neurons in major depression.


Subject(s)
Astrocytes/drug effects , Fluoxetine/pharmacology , Gene Expression Regulation/drug effects , Glucose/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction
13.
Trends Neurosci ; 34(2): 76-87, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21236501

ABSTRACT

In recent years, previously unsuspected roles of astrocytes have been revealed, largely owing to the development of new tools enabling their selective study in situ. These exciting findings add to the large body of evidence demonstrating that astrocytes play a central role in brain homeostasis, in particular via the numerous cooperative metabolic processes they establish with neurons, such as the supply of energy metabolites and neurotransmitter recycling functions. Furthermore, impairments in astrocytic function are increasingly being recognized as an important contributor to neuronal dysfunction and, in particular, neurodegenerative processes. In this review, we discuss recent evidence supporting important roles for astrocytes in neuropathological conditions such as neuroinflammation, amyotrophic lateral sclerosis and Alzheimer's disease. We also explore the potential for neuroprotective therapeutics based on the modulation of astrocytic functions.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Animals , Encephalitis/pathology , Encephalitis/physiopathology , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology
14.
J Biol Chem ; 286(10): 8585-8596, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21156804

ABSTRACT

The identification of toxic Aß species and/or the process of their formation is crucial for understanding the mechanism(s) of Aß neurotoxicity in Alzheimer disease and also for the development of effective diagnostic and therapeutic interventions. To elucidate the structural basis of Aß toxicity, we developed different procedures to isolate Aß species of defined size and morphology distribution, and we investigated their toxicity in different cell lines and primary neurons. We observed that crude Aß42 preparations, containing a monomeric and heterogeneous mixture of Aß42 oligomers, were more toxic than purified monomeric, protofibrillar fractions, or fibrils. The toxicity of protofibrils was directly linked to their interactions with monomeric Aß42 and strongly dependent on their ability to convert into amyloid fibrils. Subfractionation of protofibrils diminished their fibrillization and toxicity, whereas reintroduction of monomeric Aß42 into purified protofibril fractions restored amyloid formation and enhanced their toxicity. Selective removal of monomeric Aß42 from these preparations, using insulin-degrading enzyme, reversed the toxicity of Aß42 protofibrils. Together, our findings demonstrate that Aß42 toxicity is not linked to specific prefibrillar aggregate(s) but rather to the ability of these species to grow and undergo fibril formation, which depends on the presence of monomeric Aß42. These findings contribute significantly to the understanding of amyloid formation and toxicity in Alzheimer disease, provide novel insight into mechanisms of Aß protofibril toxicity, and important implications for designing anti-amyloid therapies.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Protein Multimerization , Alzheimer Disease/genetics , Amyloid/chemistry , Amyloid/genetics , Amyloid/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/pharmacology , Animals , Humans , PC12 Cells , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Structure, Quaternary , Rats , Rats, Sprague-Dawley
15.
J Neurochem ; 116(4): 564-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21143598

ABSTRACT

We have previously reported that the pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin-1ß (IL-1ß) induce profound modifications of the metabolic profile of astrocytes. The present study was undertaken to further characterize the effects of cytokines in astrocytes and to determine whether similar effects could also be observed in neurons. To do so, selected pro-inflammatory (IL-6 and interferon-γ, in addition to the above-mentioned TNFα and IL-1ß) and anti-inflammatory cytokines (IL-4, IL-10, transforming growth factor-ß1 and interferon-ß) were applied to primary neuronal and astrocytic cultures, and key metabolic parameters were assessed. As a general pattern, we observed that pro-inflammatory cytokines increased glucose utilization in astrocytes while the anti-inflammatory cytokines IL-4 and IL-10 decreased astrocytic glucose utilization. In contrast, no significant change could be observed in neurons. When pairs of pro-inflammatory cytokines were co-applied in astrocytes, several additive or synergistic modifications could be observed. In contrast, IL-10 partially attenuated the effects of pro-inflammatory cytokines. Finally, the modifications of the astrocytic metabolism induced by TNFα + IL-1ß and interferon-γ modulated neuronal susceptibility to an excitotoxic insult in neuron-astrocyte co-cultures. Together, these results suggest that pro- and anti-inflammatory cytokines differentially affect the metabolic profile of astrocytes, and that these changes have functional consequences for surrounding neurons.


Subject(s)
Astrocytes/metabolism , Cytokines/physiology , Inflammation Mediators/physiology , Metabolome/physiology , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/pathology , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Cytokines/classification , Down-Regulation/physiology , Energy Metabolism/physiology , Glucose/antagonists & inhibitors , Glucose/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Mice , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Up-Regulation/physiology
16.
J Cereb Blood Flow Metab ; 30(12): 1982-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20700131

ABSTRACT

Recent years have seen a surge in mathematical modeling of the various aspects of neuron-astrocyte interactions, and the field of brain energy metabolism is no exception in that regard. Despite the advent of biophysical models in the field, the long-lasting debate on the role of lactate in brain energy metabolism is still unresolved. Quite the contrary, it has been ported to the world of differential equations. Here, we summarize the present state of this discussion from the modeler's point of view and bring some crucial points to the attention of the non-mathematically proficient reader.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Blood Glucose/metabolism , Glycogen/biosynthesis , Models, Neurological
17.
J Neurosci ; 30(9): 3326-38, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20203192

ABSTRACT

Amyloid-beta (Abeta) peptides play a key role in the pathogenesis of Alzheimer's disease and exert various toxic effects on neurons; however, relatively little is known about their influence on glial cells. Astrocytes play a pivotal role in brain homeostasis, contributing to the regulation of local energy metabolism and oxidative stress defense, two aspects of importance for neuronal viability and function. In the present study, we explored the effects of Abeta peptides on glucose metabolism in cultured astrocytes. Following Abeta(25-35) exposure, we observed an increase in glucose uptake and its various metabolic fates, i.e., glycolysis (coupled to lactate release), tricarboxylic acid cycle, pentose phosphate pathway, and incorporation into glycogen. Abeta increased hydrogen peroxide production as well as glutathione release into the extracellular space without affecting intracellular glutathione content. A causal link between the effects of Abeta on glucose metabolism and its aggregation and internalization into astrocytes through binding to members of the class A scavenger receptor family could be demonstrated. Using astrocyte-neuron cocultures, we observed that the overall modifications of astrocyte metabolism induced by Abeta impair neuronal viability. The effects of the Abeta(25-35) fragment were reproduced by Abeta(1-42) but not by Abeta(1-40). Finally, the phosphoinositide 3-kinase (PI3-kinase) pathway appears to be crucial in these events since both the changes in glucose utilization and the decrease in neuronal viability are prevented by LY294002, a PI3-kinase inhibitor. This set of observations indicates that Abeta aggregation and internalization into astrocytes profoundly alter their metabolic phenotype with deleterious consequences for neuronal viability.


Subject(s)
Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Brain/metabolism , Energy Metabolism/physiology , Nerve Degeneration/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/toxicity , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/pathology , Brain/physiopathology , Cell Communication/drug effects , Cell Communication/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Energy Metabolism/drug effects , Free Radicals/metabolism , Glucose/metabolism , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Mice , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Fragments/toxicity , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors
18.
Glia ; 56(9): 975-89, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18383346

ABSTRACT

Astrocytes play an important role in nervous system homeostasis. In particular, they contribute to the regulation of local energy metabolism and to oxidative stress defence. In previous experiments, we showed that long-term treatment with interleukin 1alpha (IL-1alpha) or tumor necrosis factor-alpha (TNFalpha) alone increases glucose utilization in primary culture of mouse astrocytes. In our study, we report that a combination of IL-1beta and TNFalpha exerts a synergistic effect on glucose utilization and markedly modifies the metabolic phenotype of astrocytes. Thus, IL-1beta+TNFalpha treated astrocytes show a marked decrease in glycogen levels, a slight but not significant decrease in lactate release as well as a massive increase in both the pentose phosphate pathway and TCA cycle activities. Glutamate-stimulated glucose utilization and lactate release, a typical feature of astrocyte energy metabolism, are altered after pretreatment with IL-1beta+TNFalpha. As far as mechanisms for oxidative stress defence are concerned, we observed that treatment with IL-1beta+TNFalpha decreases cellular glutathione content and increases glutathione release into the extracellular space while stimulating superoxide anion and nitric oxide production as well as H(2)O(2) release. Interestingly, stimulation of glucose utilization by IL-1beta+TNFalpha is not affected by the antioxidant N-acetyl-L-cysteine, suggesting that cellular stress does not account for this effect. Finally, the effects of cytokines on glucose utilization appear to involve multiple signaling cascades including the phosphoinositide 3-kinase and mitogen-activated protein kinases. Taken together these results establish that a proinflammatory environment such as observed in several neuropathological conditions including Alzheimer's disease, markedly modifies the metabolic phenotype of astrocytes.


Subject(s)
Astrocytes/metabolism , Cytokines/physiology , Energy Metabolism/physiology , Inflammation Mediators/physiology , Phenotype , Animals , Astrocytes/pathology , Cells, Cultured , Glucose/metabolism , Glutamates/metabolism , Glycolysis/physiology , Mice
19.
Pharmacol Rep ; 60(6): 1001-7, 2008.
Article in English | MEDLINE | ID: mdl-19211996

ABSTRACT

Accumulating evidence supports a role for brain-derived neurotrophic factor (BDNF) in depression. However, most of these studies have been performed in animal models that have a low face validity with regard to the human disease. Here, we examined the regulation of BDNF expression in the hippocampus and amygdala of rats subjected to the chronic mild stress (CMS) model of depression, a paradigm that induces anhedonia, a core symptom of depression. We found that exposure of rats to the CMS paradigm did not modulate BDNF mRNA expression in the hippocampus and amygdala. In addition, chronic administration of imipramine, which reversed CMS-induced anhedonia, did not alter BDNF mRNA expression in these limbic structures.


Subject(s)
Amygdala/metabolism , Brain-Derived Neurotrophic Factor/genetics , Depression/metabolism , Disease Models, Animal , Gene Expression Regulation , Hippocampus/metabolism , Animals , Chronic Disease , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Stress, Psychological/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...